US20230348564A1 - Anti-cancer proteins - Google Patents

Anti-cancer proteins Download PDF

Info

Publication number
US20230348564A1
US20230348564A1 US17/923,732 US202117923732A US2023348564A1 US 20230348564 A1 US20230348564 A1 US 20230348564A1 US 202117923732 A US202117923732 A US 202117923732A US 2023348564 A1 US2023348564 A1 US 2023348564A1
Authority
US
United States
Prior art keywords
lectin
recombinant lectin
seq
cells
cancer
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US17/923,732
Other languages
English (en)
Inventor
Dhananjay Sathe
Sarvanakumar Iyappan
Dilip Pawar
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Unichem Laboratories Ltd
Original Assignee
Unichem Laboratories Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Unichem Laboratories Ltd filed Critical Unichem Laboratories Ltd
Publication of US20230348564A1 publication Critical patent/US20230348564A1/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/7056Lectin superfamily, e.g. CD23, CD72
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/168Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from plants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/37Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from fungi
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/415Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from plants
    • C07K14/42Lectins, e.g. concanavalin, phytohaemagglutinin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation

Definitions

  • the present invention relates to the use of a lectin in the treatment of cancer.
  • the present invention relates to the use of a lectin protein having an anti-angiogenic and apoptotic effects on cancer cells.
  • the active immune system is responsible for the healthy individual; as an immune system defend the several diseases or diseased conditions. It is also believed that the immune system resists even the formation of cancer by destroying cancer cell. When the immune system fails to do so, this can result in the formation of cancer.
  • cancer describes the number of diseases which are characterized by the unregulated growth and uncontrolled division of abnormal cells. Cancer can arise from virtually to any tissue or organ in the human body. Despite recent developments in medicine and the understanding of the molecular basis of cancer, the exact causes of any given type of cancer are unknown in a particular individual. Given this lack of knowledge, it remains highly difficult to find cancer treatments that would be effective for a particular individual.
  • an effective means for treating cancer may become less effective as certain types of cancers can spread from their primary source or origin. This process, called metastasis, enables cancer cells to spread to other vital parts of the body through the blood and lymph systems.
  • metastasized cells surviving in the destination tissue by overcoming local immune defenses, and acquiring their own blood supply and nutrients through the process of angiogenesis. Nevertheless, metastasis remain a key reason why effective cancer treatments are difficult to develop.
  • glycans are modified due to altered pathophysiological condition and altered glycosylation by cancer cells. These modified glycans can be easily detected by the Glycan specific binding protein or tumor specific lectin protein (hereinafter, lectin). Moreover, these proteins also play a significant role in decoding the information related to glycans. Lectins are naturally occurring carbohydrate binding proteins; which can specifically detect the cancer associated antigen due to altered glycosylation. Because of their unique ability and specificity, lectins are useful in diagnostic and therapeutic purposes.
  • Patent Number CN106397554 describes a preparation and application method to provide Cordyceps millitaris (CCM) lectin protein. Lectin-CCM and its anti-proliferation activities tested in vitro on human cervical HeLA cancer cell line.
  • CCM Cordyceps millitaris
  • Patent Number KR1020030091386 describes a process of preparation of an extract of Korean mistletoe ( Viscum album coloratura) with a lectin ingredient in an extract. When administered to a mouse experimental model, the lectin showed enhanced antitumor and anti-metastatic activity.
  • Recombinant mistletoe lectins have been used for the treatment of skin cancer, in particular malignant melanoma in the form of metastatic tumor (Stage III and Stage IV).
  • the Patent Number RU0002639445 mentions a pharmaceutical composition, containing recombinant mistletoe lectin, for treatment of melanotic cancer. Treatment with recombinant mistletoe lectins is known to extend significantly the survival rate of cancer patients.
  • U.S. Pat. No. 7,045,300 describes a lectin protein, MFA ( Maackia fauriei agglutinin) extracted from the Korean legume Maackia fauriei , its used as a diagnostic agent for cancers and therapeutically used as an anti-proliferation agent (or an anti-cancer agent) in diseases in which N-acetylneuraminic acid exists, in particular breast cancer, melanoma or hepatoma.
  • MFA Maackia fauriei agglutinin
  • U.S. patent Ser. No. 10/294,295 describes a method of treatment of cancer by modulating angiogenesis with VEGF antagonist, in particular, galectin-1 sequences have been used as VEGF antagonist to inhibit the angiogenesis for the treatment of cancer.
  • Patent Number KR1020030028855 describes an anticancer composition containing an extract of Korean mistletoe ( Viscum album coloration) with a lectin as an active ingredient for inhibition of metastases via inhibiting angiogenesis and inhibition of telomerase activity.
  • the object of the present invention is to study and develop lectins so as to make them available as anti-tumor agents.
  • a recombinant lectin for use in a method of treatment of cancer.
  • a recombinant lectin for use in a method of treatment of cancer by inhibiting angiogenesis in the cancer cell comprising administration of therapeutically effective amount of a recombinant lectin protein.
  • a recombinant lectin for use in a method of treatment of cancer by inducing apoptosis in the cancer cell comprising administration of therapeutically effective amount of a recombinant lectin protein.
  • the lectin induces early and late stage of apoptosis in the cancer cells.
  • Apotosis is the process for the programmed cell death by the signalling pathways.
  • the term “inducing apoptosis” means herein activating signalling pathways responsible for the programmed cell death of the tumor cells.
  • Metastases is the spread of the cancer cells from their primary source to other vital parts of the body through the blood and lymph systems.
  • the term “prohibiting metastases” means herein decreasing the metastases from primary source or origin of cancer by reducing the spread to the vital organs and body parts.
  • a method of treatment of cancer by inhibiting angiogenesis in the cancer cells comprising administration of therapeutically effective amount of a recombinant lectin protein to a subject.
  • a method of treatment of cancer by inducing apoptosis in the cancer cells comprises administration of therapeutically effective amount of a recombinant lectin protein to a subject.
  • a pharmaceutical composition for use in the method of treatment of cancer comprising therapeutically effective amount of a recombinant lectin protein and a pharmaceutically acceptable excipient, wherein the composition induces apoptosis in the cancer cells.
  • a method of preventing angiogenesis in tumor cells using therapeutically effective amount of a recombinant lectin protein using therapeutically effective amount of a recombinant lectin protein.
  • a method of inducing apoptosis of tumor cells using therapeutically effective amount of a recombinant lectin protein is provided.
  • the cancer is a carcinoma such as adenocarcinoma or squamous cell carcinoma.
  • the squamous cell carcinoma is the cancer of squamous cells of skin, lung, oral, thyroid, oesophagus, vaginal, cervical, ovarian, head and/or neck, prostate or bladder.
  • the effective concentration of recombinant lectin protein is from about 0.1 ⁇ g/mL to about 200 ⁇ g/mL.
  • the therapeutically effective dose of recombinant lectin protein is from about 0.1 mg/Kg to about 100 mg/Kg body weight of a subject.
  • the recombinant lectin inhibits migration and/or proliferation of endothelial cells, modulates VEGF secretion and reduces hemoglobin content and neovasculization in the cancer cells.
  • the recombinant lectin modulates one or more markers or signaling pathways selected from: ATF-2, ERK1/2; JNK; MEK-1; P90RSK; STAT-3; p53; MMPs; HGF; C-kit; Her-2; GMSCF; IL-6; IL-8; p38/MAPK; PDGF; TNFR; MPO; Galectin-3; Fol-1; CD40L; Angiopoietin-2; Kalikrein-5; Osteopontin; TNF- ⁇ ; Endoglin; MAPK/EGFR/Ras/Raf; ADBR1; CCR5; IL-4/STAT6; NF-KB; PI3K/AKT/FOXO3; PKC/CA2+; and TNF- ⁇ /JNK, TRAIL via FADD caspase-3, Leptin, Contactin-1, Notch-1 and HGFR/c-MET.
  • markers or signaling pathways selected from: ATF-2, ERK1/2;
  • the recombinant lectin is represented by an amino acid sequence having at least 60% identity to SEQ ID NO. 1 or the amino acid sequence having at least 70%, 80%, 90%, 95%, 96%, 97%, 98% or 99% homology to SEQ ID NO. 1.
  • the recombinant lectin is selected from the amino acid sequence having SEQ ID NO. 2, SEQ ID NO. 3 or SEQ ID NO. 4.
  • the recombinant lectin is a modified lectin protein (i.e. a recombinant lectin protein having at least one amino acid modification in a carbohydrate binding site) as defined in WO2020/044296 which is incorporated herein by reference, in particular with regard to the definition of the lectin.
  • the recombinant lectin comprises at least one amino acid modification in a carbohydrate binding site of SEQ ID NO. 1 or an amino acid sequence having at least 60% homology to SEQ ID NO. 1.
  • the carbohydrate binding site is a primary and/or secondary carbohydrate binding site.
  • the primary carbohydrate binding site comprises a position selected from 1 or more of 27, 28, 47, 48, 70, 71, 72 & 105 in SEQ ID NO. 1 or in an amino acid sequence having at least 60% homology to SEQ ID NO. 1.
  • the position of the amino acid modification is selected from one or more of:
  • the secondary carbohydrate binding site comprises a position selected from one or more of 77, 78, 80, 101, 112, and 114 in SEQ ID NO. 1 or in an amino acid sequence having at least 60% homology to SEQ ID NO. 1.
  • the position of the amino acid modification is selected from one or more of:
  • the amino acid modification is an amino acid substitution such that a substituting amino acid replaces an original amino acid.
  • amino acid substitution in the primary carbohydrate binding site is selected from one or more of:
  • amino acid substitution in the secondary carbohydrate binding site is selected from one or more of:
  • the lectin protein comprises at least one amino acid modification in the N-terminus of SEQ ID NO.1 or in an amino acid sequence having at least 60% homology to SEQ ID NO. 1, wherein the N-terminus comprises a position selected from: 1 and/or 2 in SEQ ID NO. 1 or a corresponding position in the sequence having at least 60%, 70%, 80%, 90%, 95%, 97% or 99% homology thereto.
  • the amino acid modification is an amino acid substitution at position 1 and wherein a substituting amino acid is not threonine or valine.
  • the substituting amino acid is selected from: alanine, glycine, proline or serine.
  • the amino acid modification is an amino acid substitution at position 2 and wherein a substituting amino acid is tryptophan.
  • cleavage of an initiator methionine is increased or decreased as compared with a control.
  • the amino acid modification at position 76 is an amino acid substitution with a non-polar amino acid.
  • the non-polar amino acid is selected from: glycine, valine or leucine.
  • the amino acid modification at position 44 or 89 is an amino acid substitution with a non-polar amino acid.
  • non-polar amino acid is selected from: leucine, isoleucine or valine.
  • the modified lectin protein is soluble, partially soluble or insoluble and/or has cytotoxicity.
  • the modified lectin protein has a cytotoxicity that is at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of a control.
  • the modified lectin protein has a percentage cytotoxicity that is less than 10% of a control, or is absent of cytotoxicity.
  • the modified lectin protein is equal to or less than 500, 400, 300, 250, 200, or 150 amino acids in length.
  • the present invention provides an effective anti-angiogenesis using from about 0.1 mg/Kg to 100 mg/Kg body weight of a recombinant lectin having the amino acid sequence of SEQ ID NO.2.
  • the present invention further relates to effective anti-angiogenesis in tumor cells using from about 0.1 ⁇ g/mL to 200 ⁇ g/mL concentration of a recombinant lectin having the amino acid sequence of SEQ ID NO. 2.
  • the present invention provides a method of treatment of adenocarcinoma, squamous cell carcinoma and/or brain cancer by preventing angiogenesis and/or by inducing apoptosis in tumor cells using a recombinant lectin having the amino acid sequence of SEQ ID NO.2.
  • the present invention provides a method of treatment of adenocarcinoma, squamous cell carcinoma and/or brain cancer by preventing angiogenesis and/or by inducing apoptosis using from about 0.1 mg/Kg to 100 mg/Kg body weight of a recombinant lectin having the amino acid sequence of SEQ ID NO. 2.
  • the present invention provides a method of treatment of adenocarcinoma, squamous cell carcinoma and/or brain cancer by preventing angiogenesis and/or by inducing apoptosis in the tumor cells using from about 0.1 ⁇ g/mL to about 200 ⁇ g/mL concentration of a recombinant lectin having the amino acid sequence of SEQ ID NO.2.
  • the present invention further relates to the evaluation of the modulatory effect of a recombinant lectin having the amino acid sequence of SEQ ID NO.2 on key signaling pathways involved in the pathogenesis of cancer.
  • SEQ ID NO: 4 represents a variant of the S. rolfsii lectin amino acid sequence (reported in WO 2014/203261).
  • amino acids can be grouped according to different biochemical properties. Examples include: the polar amino acids, the non-polar amino acids, the acidic amino acids and the basic amino acids.
  • the amino acid used for the amino acid modification is at least one selected from the group consisting of, but not limited to: polar, non-polar, acidic, basic, selenocysteine, pyrrolysine and non-canonical.
  • homology refers to two or more referenced entities that share at least partial identity over a given region or portion. Areas, regions or domains of homology or identity refer to a portion of two or more referenced entities that share homology or are the same. Thus, where two sequences are identical over one or more sequence regions they share identity in these regions.
  • Substantial homology refers to a molecule that is structurally or functionally conserved such that it has or is predicted to have at least partial structure or function of one or more of the structures or functions (e.g., a biological function or activity) of the reference molecule, or a relevant/corresponding region or portion of the reference molecule to which it shares homology.
  • the percentage “homology” between two sequences is determined using the BLASTP algorithm with default parameters (Altschul et al. Nucleic Acids Res. 1997 Sep. 1; 25(17):3389-402).
  • the BLAST algorithm can be accessed on the internet using the URL: https://blast.ncbi.nlm.nih.gov/Blast.cgi.
  • percentage homology between two sequences is determined using the EMBOSS Needle algorithm using default parameters.
  • the EMBOSS Needle algorithm can be accessed on the internet using the URL: https://www.ebi.ac.uk/Tools/psa/emboss_needle/.
  • sequence identity is used interchangeably with the term “sequence identity” in the present specification.
  • recombinant protein or “recombinant polypeptide” as used herein refers to a protein molecule which is expressed using a recombinant DNA molecule.
  • the recombinant protein according to present invention is the protein having amino acid sequence of SEQ ID 1 which is also referred as SEQ ID 1.
  • recombinant protein is intended here to cover any pharmaceutically acceptable salt, solvate, hydrate, prodrug, or any other compound which, upon administration to the patient is capable of providing (directly or indirectly) the compound as described herein.
  • the preparation of salts, solvates, hydrates, and prodrugs can be carried out by methods known in the art.
  • an inventive combination may vary depending on such factors as the desired biological endpoint, the pharmacokinetics of the agents being delivered, the disease being treated, the mode of administration, and the patient. Treatment is generally “effective” if one or more symptoms or clinical markers are reduced. Alternatively, treatment is “effective” if the progression of a disease, disorder or medical condition is reduced or halted.
  • terapéuticaally effective amount is an amount sufficient to effect desired clinical results (i.e., achieve therapeutic efficacy).
  • a therapeutically effective amount can be administered in one or more administrations.
  • a therapeutically effective amount of a recombinant protein is an amount that is sufficient to palliate, ameliorate, stabilize, reverse, prevent, slow or delay the progression of the disease state.
  • angiogenesis is prevented/inhibited in a tumour, such as a tumour in a mammalian body, e.g. a human body.
  • the lectin is derived from a soil borne phytopathogenic fungus, such as S. rolfsii .
  • a soil borne phytopathogenic fungus such as S. rolfsii .
  • the lectin comprises an amino acid sequence which is identical or similar to a native sequence and is synthesized in the laboratory using recombinant DNA technology.
  • the lectin may comprise an amino acid sequence having at least 60% 70%, 80%, 90%, 95%, 96%, 97%, 98% or 99% homology to a native sequence.
  • the host cell can then be cultured under appropriate conditions, whereby the recombinant lectin is expressed.
  • the recombinant lectin can thus be obtained by isolation as an expression product from the host cell.
  • Recombinant proteins can be purified by conventional techniques known in the art, typically conventional chromatographic methods.
  • the lectin is specific for the TF antigen. In some embodiments the lectin is specific for O-glycans.
  • the lectin has an IC 50 value of no more than 20 ⁇ g/mL against a cervical carcinoma cell line, such as the human KB cell line.
  • the lectin has an IC 50 value of no more than 10 ⁇ g/mL against a breast cancer cell line (e.g. mammary gland adenocarcinoma, breast adenocarcinoma or breast metastatic carcinoma), such as the human MDA-MB-231 cell line.
  • a breast cancer cell line e.g. mammary gland adenocarcinoma, breast adenocarcinoma or breast metastatic carcinoma
  • the lectin has an IC 50 value of no more than 15 ⁇ g/mL against a bladder cancer cell line (e.g. urinary bladder carcinoma or transitional cell carcinoma), such as the human T-24 cell line.
  • a bladder cancer cell line e.g. urinary bladder carcinoma or transitional cell carcinoma
  • the lectin has an IC 50 value of no more than 15 ⁇ g/mL and 20 ⁇ g/mL against a Brain tumour cell lines such as U251MG (Glioblastoma) and IOMM-Lee (Meningioma), respectively.
  • a Brain tumour cell lines such as U251MG (Glioblastoma) and IOMM-Lee (Meningioma), respectively.
  • IC 50 values for a given therapeutic agent can be determined using standard techniques as would be known by the skilled person.
  • the IC 50 value of a lectin for a particular type of cancer may be determined in vitro using a suitable cell line which is representative of that type of cancer.
  • the cell line may be treated with the lectin protein, optionally along with a control agent, which is an established anti-cancer agent.
  • the cell cytotoxicity may be estimated in an untreated sample, the test sample and the control using processes well known to the person skilled in the art, which may involve Calcein AM Cell Viability assay or MTT assay or any other method known to the skilled person. Percentage cytotoxicity with respect to untreated cells may be calculated using formula:
  • the IC 50 value may be calculated using software know to the skilled person, such as Pad Prism version 4.01 software.
  • the lectin may be provided in a pharmaceutically acceptable form, such as a liquid (e.g in an aqueous solution or suspension, or as an oil based solution or suspension.), a solid (e.g a capsule or tablet), a lyophilized powder, a spray, cream, lotion or gel, vesicular drug delivery systems such as, but not limited to, bilosomes, liposomes, niosomes, transferosome, ethosomes, sphingosomes, pharmacosomes, multilamellar vesicles, microsphere and the like.
  • a pharmaceutically acceptable form such as a liquid (e.g in an aqueous solution or suspension, or as an oil based solution or suspension.), a solid (e.g a capsule or tablet), a lyophilized powder, a spray, cream, lotion or gel, vesicular drug delivery systems such as, but not limited to, bilosomes, liposomes, niosomes, transfero
  • cancer tumors
  • tumors result from abnormal cell growth. They form when the normal cells grow out of control and crowd out. Formation of tumours often affects the normal functioning of the tissue, organ or organism.
  • cancer encompasses both primary and metastatic cancers.
  • cancer includes, but is not limited to, solid tumors and blood borne tumors.
  • cancer includes diseases of the skin, tissues, organs, bone, cartilage.
  • examples of cancers that may be treated by the methods and compositions of the present invention include, but are not limited to, cancer of the bile duct, bladder, bone, brain, breast, cervix, colon, oesophagus, gastrointestine (including the ileum, colon, rectum and/or anus), head, kidney, liver, lung, nasopharynx, neck, ovary, pancreas, prostate, skin, stomach, testis, tongue, thyroid, urachus, vagina & uterus.
  • the cancer may be benign or malignant, and in any stage of malignancy.
  • the cancer may be a cancer of the epithelial tissues, non-epithelial tissues, the cells that make up the skin or the tissue lining the organs, cells of the immune system, connective tissue, or cells of the spinal cord or brain.
  • the cancer may be a solid tumour.
  • the cancer may be a carcinoma.
  • the cancer is adenocarcinoma.
  • the adenocarcinoma may be oesophageal, pancreatic, prostate, cervical, breast, colon or colorectal, lung, bile duct, vaginal, urachus or stomach adenocarcinoma.
  • the cancer is squamous cell carcinoma.
  • the squamous cell carcinoma may be skin, oral, lung, thyroid, oesophagus, vaginal, cervical, ovarian, head and/or neck, prostate or bladder squamous cell carcinoma.
  • the cancer may be brain tumor/cancer, which might include Glioblastoma, meningioma, astrocytoma, glioma and neuroblastoma.
  • treatment may comprise substantially curing the cancer, preventing or slowing the progression of, or reducing the severity of, the disease, preventing or reducing metastases, inhibiting tumour growth, reducing tumour mass or eliminating tumours, and/or ameliorating (either temporarily or permanently) symptoms associated with the disease. It will be appreciated that symptoms will vary depending on the type of cancer, but may include pain, reduction or loss of function, nausea and/or sickness, fever, tumour formation, immunosuppression, and/or tiredness.
  • the treatment may comprise administering a therapeutically effective amount of the lectin to the subject.
  • the lectin is administered at a dose of from about 0.05 mg/Kg to about 1000 mg/Kg, from about 0.1 mg/Kg to about 100 mg/Kg.
  • the treatment comprises administering the lectin to a subject such that the effective concentration of the lectin in the subject is from about 0.001 ⁇ g/mL to about 1000 ⁇ g/mL, 0.05 ⁇ g/mL to about 500 ⁇ g/mL, from 0.1 ⁇ g/mL to 200 ⁇ g/mL, from 0.15 ⁇ g/mL to 150 ⁇ g/mL.
  • the cancer is selected from breast cancer (e.g. breast adenocarcinoma), cervical carcinoma, ovarian cancer (e.g. ovarian squamous cell carcinoma) and pancreatic cancer (e.g. pancreatic adenocarcinoma), bladder cancer (e.g urothelial carcinoma, urothelial carcinoma), Brain cancer (e.g. Glioblastoma, meningioma, astrocytoma, glioma and neuroblastoma) and the treatment comprises administering the lectin to the subject such that the effect concentration of the lectin in the subject is from 0.1 ⁇ g/mL to 200 ⁇ g/mL.
  • breast cancer e.g. breast adenocarcinoma
  • cervical carcinoma e.g. ovarian squamous cell carcinoma
  • pancreatic cancer e.g. pancreatic adenocarcinoma
  • bladder cancer e.g. pancreatic adenocarcinoma
  • the treatment comprises administering a non-cytotoxic concentration of the lectin.
  • Administration of the lectin may be by any suitable route, including but not limited to, injection (including intravenous (bolus or infusion), intra-arterial, intraperitoneal, subcutaneous (bolus or infusion), intraventricular, intramuscular, or subarachnoidal), oral ingestion (e.g. of a tablet, gel, lozenge or liquid), inhalation, topical, via a mucosa (such as the oral, nasal or rectal mucosa), by delivery in the form of a spray, tablet, transdermal patch, subcutaneous implant or in the form of a suppository.
  • injection including intravenous (bolus or infusion), intra-arterial, intraperitoneal, subcutaneous (bolus or infusion), intraventricular, intramuscular, or subarachnoidal
  • oral ingestion e.g. of a tablet, gel, lozenge or liquid
  • inhalation e.g. of a tablet, gel, lozenge or liquid
  • topical e.g. of a tablet, gel
  • the subject may be a mammalian subject. In some embodiments, the subject is human.
  • Angiogenesis plays an important role in the growth and progression of cancer. Blood vessels penetrating into the tumour parenchyma provide nutrition and oxygen for multiplying cells.
  • the control of tumour angiogenesis depends on a net balance of several activators (angiogenic factors) and inhibitors (anti-angiogenic factors), which are secreted by both tumour cells and host infiltrating cells such as macrophages and fibroblasts.
  • the angiogenic factors induce endothelial cells to secrete proteases and plasminogen activators that degrade the vessel basement membrane, leading to cell invasion into the surrounding matrix and the formation of new vessels.
  • Potent anti-angiogenic molecules inhibit the proliferation and migration of endothelial cells, by binding to the pro-angiogenic factors or blocking the activities of receptors on the surface of endothelial cells.
  • the lectin is capable of inhibiting the migration and/or proliferation of cells, such as endothelial cells.
  • a lectin to inhibit the migration and/or proliferation of cells can be tested using standard techniques, such as those described herein.
  • Non-cytotoxic concentrations of recombinant lectin having the amino acid sequence of SEQ ID NO. 2 demonstrating anti-proliferative and hence anti-angiogenic effects in endothelial cells may be from 10 ⁇ g/mL to 100 ⁇ g/mL.
  • the recombinant lectins of the present invention such as having the amino acid sequence of SEQ ID NO. 2 showed dose dependent inhibitory effect on serum mediated cell proliferation.
  • the non-cytotoxic concentrations of the recombinant lectin having the amino acid sequence of SEQ ID NO. 2 demonstrated anti-proliferative and hence anti-angiogenic effects in endothelial cells, at concentrations which ranged from about 20 ⁇ g/mL to 100 ⁇ g/mL.
  • the determination of the anti-angiogenic effect of a recombinant lectin having the amino acid sequence of SEQ ID NO.2 in human endothelial cells EA.Hy926 was performed with a concentration of lectin ranging from about 0.1 ⁇ g/mL to 200 ⁇ g/mL.
  • a concentration of from 20 ⁇ g/mL to 100 ⁇ g/mL resulted in the inhibition of endothelial cell proliferation by from 15.51% to 58.53% as compared to control.
  • the same concentration range resulted in the inhibition of endothelial cells migration by from 71.5% to 82.4% as compared to control (DMEM) after 72 hrs.
  • the present invention further relates to the evaluation of in-vivo anti-angiogenesis potential of recombinant lectin (such as recombinant lectin having the amino acid sequence of SEQ ID NO. 2 using Matrigel plug assay in C57BL/6 mice.
  • recombinant lectin such as recombinant lectin having the amino acid sequence of SEQ ID NO. 2
  • the group of mice treated with recombinant lectin having the amino acid sequence of SEQ ID NO. 2 showed 23.6% reduction in haemoglobin content in homogenate of Matrigel plug, whereas the group treated with Sunitinib showed maximum reduction i.e. 59.2% in haemoglobin content in homogenate of Matrigel plug.
  • Further neovascularization was marginally reduced in mice treated with Recombinant Lectin having the amino acid sequence of SEQ ID No 2 at 10 mg/Kg.
  • the present invention further relates to the evaluation of modulatory effect of recombinant lectins (such as a recombinant lectin having the amino acid sequence of SEQ ID No 2) on signaling pathways involved in pathogenesis of cancer.
  • recombinant lectins such as a recombinant lectin having the amino acid sequence of SEQ ID No 2
  • the lectin may modulate one or more biomarkers selected from: MEK-1; P90RSK; STAT-3; p53; MMPs; HGF; EGF; C-kit; VEGF; VEGFR; Her-2/3; GMSCF; IL-6; IL-8; p38/MAPK; PDGF; MPO; Fol-1; CD40L; Angiopoietin-2; Osteopontin; Endoglin; P1GF; BMP-9; Endothelin-1.
  • biomarkers selected from: MEK-1; P90RSK; STAT-3; p53; MMPs; HGF; EGF; C-kit; VEGF; VEGFR; Her-2/3; GMSCF; IL-6; IL-8; p38/MAPK; PDGF; MPO; Fol-1; CD40L; Angiopoietin-2; Osteopontin; Endoglin; P1GF; BMP-9; Endothelin-1.
  • the lectin modulates one or more biomarkers or signaling pathways selected from MAPK/EGFR/Ras/Raf; ADBR1; CCR5; NF-KB; PI3K/AKT/FOXO3; and PKC/CA2+.
  • the inhibiting concentration range of the recombinant lectin for the MAPK/EGFR/Ras/Raf and ADBR1 pathways is between 0.158 ⁇ g/mL to 50 ⁇ g/mL with an effective inhibition from 2% to 48% and from 26% to 49%, respectively for the two pathways.
  • the inhibiting concentration range of the recombinant lectin for the NF-KB, TNF-alpha/JNK and PI3K/AKT/FOXO3 pathways is between 0.5 ⁇ g/mL to 50 ⁇ g/mL with an effective inhibition from 3% to 13%, from 12% to 45% and from 2% to 73%, respectively for the 3 pathways.
  • the inhibiting concentration range of the recombinant lectin for the CCR5 pathway is in the range of 0.058 ⁇ g/mL to 50 ⁇ g/mL with effective inhibition of 21%-70%.
  • the inhibiting concentration range of the recombinant lectin for the PKC/Ca2+ pathway in the range of 0.00158 ⁇ g/mL to 0.5 ⁇ g/mL with an effective inhibition from 5% to 19%.
  • the lectin modulates VEGF levels. It will be understood that, as used herein, the term “modulates” refers to the ability of an agent to increase or decrease the expression or activity level of a biomarker or signalling pathway, compared to normal levels (i.e. in untreated cells). In some embodiments, the lectin increases expression of VEGF in cells, such as cancer cells.
  • VEGF Vascular epidermal growth factor
  • PSI proteasome inhibitor
  • the present inventors have found that the treatment of cancer cells with a lectin in accordance with the present invention resulted in an increase in VEGF levels, as compared to untreated cells. Without being bound by theory, it is thought that the lectins of the present invention may exert their anti-cancer effect in a similar way to PSI, by increasing the susceptibility of endothelial cells to the pro-apoptitic activity of the lectin, thereby exerting an anti-angiogenic effect.
  • the lectin further induces apoptosis of cancer cells.
  • the anti-angiogenic effect of the lectin may be determined by a reduction of tumor mass or volume, by the percentage inhibition of tumour growth (% TGI) or by the disappearance of tumours. In some embodiments, the anti-angiogenic effect of the lectin may be determined by an increase in the time required for a tumour to reach a predetermined mass or volume, as compared to an untreated control.
  • the lectin inhibits (or is capable of inhibiting) tumour growth by at least 20%, 30%, 35%, 40%, 45%, 50%, 55%, or at least 60%.
  • the % tumour growth inhibition (% TGI) can be determined using methods described herein.
  • the lectin effects (or is capable of effecting) a tumour growth delay of at least 2, 3, 4, 5, 6, 7, 8, 10, 12 or at least 14 days.
  • the Tumour Growth Delay can be determined using methods described herein.
  • a method of treating cancer in a subject comprising administering a lectin to the subject, wherein the lectin effects treatment of the cancer by inhibiting angiogenesis.
  • the present invention further relates to a method of preventing angiogenesis in a tumor in a subject, using a lectin in accordance with the present invention.
  • the method of preventing angiogenesis in the tumor comprises using a non-cytotoxic concentration of the lectin, such as a recombinant lectin having the amino acid sequence of SEQ ID NO. 1 or its homologous sequence.
  • the non-cytotoxic concentration of the lectin may be from about 0.1 ⁇ s/mL to about 200 ⁇ g/mL.
  • the method may comprise contacting a solution of the lectin (e.g. solution of recombinant lectin having the amino acid sequence of SEQ ID NO.1 or its homologous sequence, at a concentration of about 0.1 ⁇ g/mL to about 200 ⁇ g/mL) with tumor cells.
  • the present invention relates to effecting anti-angiogenesis in tumor cells using from about 0.1 mg/Kg to 100 mg/Kg body weight of a lectin, such as a recombinant lectin comprising the amino acid sequence of SEQ ID NO.1 or its homologous sequence.
  • a lectin such as a recombinant lectin comprising the amino acid sequence of SEQ ID NO.1 or its homologous sequence.
  • references to mg/Kg of body weight refer to mammalian body weight, such as human body weight.
  • recombinant lectin having amino acid sequence of SEQ ID 1 or its homologous sequence with concentration of about 0.1 ⁇ g/mL to about 200 ⁇ g/mL, is contacted with the tumor cells of mammalian body with amount from about 0.1 mg/Kg to 100 mg/Kg of the mammalian body weight for effective anti-angiogenesis effect in the tumor cells.
  • a lectin for use in a method of treatment of cancer by inducing apoptosis.
  • the invention provides a recombinant lectin for use in a method of treatment of cancer by inducing apoptosis in the cancer cell comprising administration of therapeutically effective amount of a recombinant lectin protein.
  • the lectin induces early and late stage of apoptosis in the cancer cells.
  • the present invention further relates to a method of inducing apoptosis in a tumor in a subject, using a recombinant lectin in accordance with the present invention.
  • the present invention also relates to the evaluation of in vitro apoptotic effect of a lectin (such as a recombinant lectin having the amino acid sequence of SEQ ID NO. 1 or SEQ ID NO. 2) on a cancer cell line, for example on a breast cancer cell line and/or a pancreatic cancer cell line.
  • a lectin such as a recombinant lectin having the amino acid sequence of SEQ ID NO. 1 or SEQ ID NO. 2
  • the in vitro apoptotic effect of lectins may be determined using standard assay techniques which are known to the skilled person.
  • the evaluation of the in vitro apoptotic effect of a recombinant lectin having the amino acid sequence of SEQ ID NO. 2 on MDA-MB-231 as a breast cancer cell line and PANC-1 cell line as a pancreatic cancer cell line was carried out using a JC-1 assay.
  • the evaluation was performed using a concentration of recombinant lectin of about 2.5 ⁇ g/mL-80 ⁇ g/mL, with Doxorubicin as a positive control.
  • the recombinant lectin having the amino acid sequence of SEQ ID NO. 2 led to a significant depolarization of mitochondrial membrane in the range of 9.5%-51.7% for PANC-1 cell line and 19.8%-54.1% for MDA-MB-231 cell line.
  • the present invention further relates to the evaluation of modulatory effect of recombinant lectins (such as a recombinant lectin having the amino acid sequence of SEQ ID NO.2) on signaling pathways involved in pathogenesis of cancer.
  • recombinant lectins such as a recombinant lectin having the amino acid sequence of SEQ ID NO.2
  • SEQ ID NO. 2 induced apoptosis by modulation of one or more biomarkers selected from: MEK-1; P90RSK; STAT-3; p53; C-kit; IL-6; IL-8; p38/MAPK; MPO; Fol-1; CD40L; ATF-2, ERK1/2; JNK; TNFR; Galectin-3; Kalikrein-5 and TNF- ⁇ .
  • biomarkers selected from: MEK-1; P90RSK; STAT-3; p53; C-kit; IL-6; IL-8; p38/MAPK; MPO; Fol-1; CD40L; ATF-2, ERK1/2; JNK; TNFR; Galectin-3; Kalikrein-5 and TNF- ⁇ .
  • SEQ ID NO. 2 modulates one or more biomarkers or signaling pathways selected from IL-4/STAT6; NF-KB; PI3K/AKT/FOXO3; and TNF- ⁇ /JNK.
  • the inhibiting concentration range of the recombinant lectin for the NF-KB, PI3K/AKT/FOXO3 and TNF-alpha/JNK pathways is between 0.5 ⁇ g/mL to 50 ⁇ g/mL with an effective inhibition from 3% to 13%, from 2% to 73% and from 12% to 45%, respectively for the 3 pathways.
  • the inhibiting concentration range of the recombinant lectin for the IL-4/STAT6 pathway is in the range of 0.0158 ⁇ g/mL to 0.5 ⁇ g/mL with an effective inhibition from 16% to 28%.
  • the method of inducing apoptosis in the tumor comprises using a non-cytotoxic concentration of the lectin, such as a recombinant lectin having the amino acid sequence of SEQ ID NO. 1 or homologous sequence thereof.
  • the non-cytotoxic concentration of the lectin may be from about 0.1 ⁇ g/mL to about 200 ⁇ g/mL.
  • the method may comprise contacting the recombinant lectin or the solution of recombinant lectin having the amino acid sequence of SEQ ID NO.1 or its homologous sequence, at a concentration of about 0.1 ⁇ g/mL to about 200 ⁇ g/mL with tumor cells.
  • the present invention relates to effecting apoptosis in tumor cells using from about 0.1 mg/Kg to 100 mg/Kg body weight of a recombinant lectin, such as a recombinant lectin comprising the amino acid sequence of SEQ ID NO.1 or its homologous sequence.
  • the treatment of the cancer or the effective apoptosis of the tumor cells may be determined by a reduction in tumour volume or by the disappearance of one or more tumours.
  • the recombinant lectin comprises or consists of the amino acid sequence of SEQ ID No 1 or SEQ ID No 2 or SEQ ID No 3, or SEQ ID No 4.
  • the method of treatment of cancer, the method of preventing angiogenesis, or the method of inducing apoptosis may comprise contacting the tumour with a composition comprising the recombinant lectin.
  • a composition comprising the recombinant lectin.
  • a solution of recombinant lectin having amino acid sequence of SEQ ID NO.1 or its homologous sequence may be contacted with the tumour cells.
  • the concentration of the recombinant lectin in the composition may be from about 0.001 ⁇ g/mL to about 1000 ⁇ g/mL, about 0.05 ⁇ g/mL to about 500 ⁇ g/mL, from about 0.1 ⁇ g/mL to about 200 ⁇ g/mL, or from about 0.15 ⁇ g/mL to about 150 ⁇ g/mL.
  • the method of treatment of cancer, the method of preventing angiogenesis or the method of inducing apoptosis may comprise administering the recombinant lectin in an amount of from about 0.05 mg/Kg to about 1000 mg/Kg, or from about 0.1 mg/Kg to about 100 mg/Kg of a mammalian body weight.
  • a method of treatment of adenocarcinoma or squamous carcinoma or brain cancer by preventing angiogenesis and/or by inducing apoptosis in tumour cells comprising contacting the tumour cells with a solution of a recombinant lectin, such as a recombinant lectin having the amino acid sequence of SEQ ID NO. 1 or its homologous sequence.
  • concentration of the solution of the recombinant lectin e.g. SEQ ID NO.1, or SEQ ID NO. 2
  • concentration of the solution of the recombinant lectin may be is from about 0.1 ⁇ g/mL to 200 ⁇ g/mL.
  • the present invention further relates to the evaluation of anti-tumor potential of recombinant lectins, such as a recombinant lectin having the amino acid sequence of SEQ ID NO. 2.
  • the anti-tumor potential of a recombinant lectin having the amino acid sequence of SEQ ID NO. 2 was evaluated using a PA-1 (ovary teratocarcinoma) xenograft model. Nude mice treated with recombinant Lectin showed significant decrease in tumor size upon treatment.
  • the anti-tumor potential of the recombinant lectin was also evaluated using a KB (cervical carcinoma) xenograft model.
  • the percentage tumour growth inhibition was comparable to the standard (Doxorubicin).
  • the anti-tumor potential of the recombinant lectin was further evaluated using a HT-29 (colorectal adenocarcinoma) xenograft model. Animals treated with the recombinant lectin at 20 mg/Kg and 30 mg/Kg body weight (daily) showed a significant decrease in tumor volume in comparison with the vehicle control group. Animals were further treated with the recombinant lectin using a T24 (urinary bladder/transitional cell carcinoma) Xenograft Model. The Recombinant Lectin showed appreciable anti-tumor potential which was comparable to a Doxorubicin-treated group in the T24 xenograft model.
  • the anti-tumor potential of the recombinant lectin was further evaluated using breast cancer cell lines (MCF-7 and MDA-MB-231).
  • the percentage tumour growth inhibition was comparable to the standard (Doxorubicin) in both the cases.
  • PANC-1 Pancreatic/duct epithelioid carcinoma
  • the present invention further relates to a recombinant lectin (e.g. a recombinant lectin having the amino acid sequence of SEQ ID NO.2) for use as an angiogenesis inhibitor or apoptosis inducer, thereby inhibiting cancer cell metastases and/or causing programmed cell death.
  • a recombinant lectin e.g. a recombinant lectin having the amino acid sequence of SEQ ID NO.2
  • the purified recombinant lectin of SEQ ID No 2 was studied for its anti-cancer potential in different cell lines. It has shown the cytotoxic activity in the 10 different cancer cell lines.
  • the brief method of the assay was as follows:
  • the recombinant lectin having amino acid sequence of SEQ ID No 2 showed cytotoxic effect on all 10 cancer cell lines tested and did not show cytotoxic effect on the normal cells (PBMCs). It showed better effect in MDA-MB-231 (triple negative breast adenocarcinoma cells) compared to MCF-7 (Breast adenocarcinoma). The results are summarized in table below.
  • the recombinant lectin of SEQ ID No 2 exhibited cytotoxic anti-proliferation effect on various cancer cell lines in in vitro assays.
  • the efficacy of the recombinant lectin having amino acid sequence of SEQ ID No 2 as antitumor agent was assessed in respective Xenograft in immunocompromised mice models in vivo.
  • the Xenograft models used were HT-29, KB, PA-1, MCF-7, PANC-1, T24 and MDA-MB-231.
  • the basic study design for the Xenograft study was as follows:
  • the mechanism of action of the recombinant lectin having the amino acid sequence of SEQ ID NO.2 is studied by determining its effect on the modulation of key signaling pathways involved in the pathogenesis of cancer. The study was performed using SelectScreen® Cell-based Pathway Profiling Services at Life Technologies, USA. Modulatory effect of Lectin having the amino acid sequence of SEQ ID NO. 2 on various cell signaling pathways was investigated in cell lines overexpressing specific markers using GeneBLAzer Beta-lactamase (bla) Reporter Technology and Tango platforms.
  • the cell lines tested were MDA-MB-231 (Human Breast cancer), KB (Human Cervical cancer), PA-1 (Human ovarian cancer), PANC-1 (Human pancreatic cancer), HT-29 (Human colorectal cancer), T-24 (Human Bladder cancer)
  • recombinant lectin having the amino acid sequence of SEQ ID NO.2 was dissolved in 200 ⁇ L of TBS buffer (25 mM, pH 8.0) to obtain stock solution of 50 mg/mL.
  • the stock solutions were diluted in Serum Free Medium (STM) to achieve final concentrations in cells ranging from 0.00158 ⁇ g/mL-50 ⁇ g/mL.
  • Cells (32 ⁇ L) were diluted in Assay Media to appropriate cell density and were added to the assay plate. Cells were incubated at 37° C./5% CO2 for 24 h.
  • A Fluorescence reading in Control (untreated cells)
  • B Fluorescence reading in TI treated cells
  • the cell lines MDA-MB-231 Human breast adenocarcinoma
  • PANC-1 Human pancreatic epitheloid carcinoma
  • DMEM Dulbecco's Modified Eagle Medium
  • the cell line was subcultured by trypsinization followed by splitting the cell suspension into fresh flasks and supplementing with fresh culture medium.
  • Recombinant lectin having the amino acid sequence of SEQ ID NO. 2 was diluted in serum free medium.
  • Doxorubicin was used as positive control and stock solution was prepared in dimethyl sulfoxide (DMSO).
  • the MDA-MB-231 and PANC-1 cells were trypsinized, counted and plated in wells of flat bottom 96-well plate (dark walled plate) at the density corresponding to 10 ⁇ 10 3 cells/well/180 ⁇ l DMEM with 10% FBS. The cells were then incubated overnight under growth conditions to allow the cell recovery and exponential growth. Cells were treated with the recombinant lectin of SEQ ID NO. 2 (20 ⁇ l stock solution) to achieve final concentrations of 2.5 ⁇ g/mL, 5 ⁇ g/mL, 10 ⁇ g/mL, 20 ⁇ g/mL, 40 ⁇ g/mL and 80 ⁇ g/mL.
  • cells were treated with doxorubicin to achieve final concentrations of 0.1 ⁇ M, 1 ⁇ M, 10 ⁇ M, 25 ⁇ M and 50 ⁇ M. Following respective treatments the cells in the above plate were incubated for 16h in CO2 incubator at 37° C., 5% CO2 and 95% humidity.
  • JC1-dye solution prepared by diluting 1 mM DMSO stock in to 10 ⁇ M in 1 ⁇ PBS
  • the cells were then incubated with the dye in CO2 incubator at 37° C. for 15 min. After 15 min of incubation, the supernatant was removed and the cells were washed twice with 1 ⁇ PBS and then 100 ⁇ L of 1 ⁇ PBS was added to each well.
  • Red fluorescence excitation 550 nm, emission 600 nm
  • green fluorescence excitation 485 nm, emission 535 nm
  • the mitochondrial membrane potential (ATM) was calculated as the ratio of intensity of red fluorescence to intensity of green fluorescence described as follows:
  • ⁇ M Intensity of red fluorescence/Intensity of green fluorescence
  • MMP mitochondrial membrane potential
  • Cell lines MDA-MB-231 Human breast adenocarcinoma
  • PANC-1 Human pancreatic epitheloid carcinoma
  • the cell lines were maintained in DMEM+10% FBS (heat inactivated) at 37° C. with 5% CO2, and 95% humidity.
  • Antibiotics Penicillin (100 U/mL) and Streptomycin (100 ⁇ g/mL) were added to the medium.
  • the cell lines were subcultured by trypsinization, followed by splitting the cell suspension into fresh flasks and supplementing with fresh culture medium.
  • the stock solution of recombinant lectin having the amino acid sequence of SEQ ID NO. 2 was diluted in Serum Free Medium (SFM) at different concentrations corresponding to 10 fold high concentrations (weight by volume) of final effective concentration.
  • SFM Serum Free Medium
  • Doxorubicin was used as positive control and stock solution was prepared in DMSO.
  • Cells were counted using hemocytometer and plated in culture plates at the density of 0.4 ⁇ 10 6 cells/well in DMEM+10% FBS in 6-well plates. Cells were incubated overnight to allow the cell recovery and exponential growth. Following overnight incubation, cells were treated with Lectin having the amino acid sequence of SEQ ID NO. 2 in DMEM+0% FBS at concentrations ranging from 2.5 ⁇ g/mL to 80 ⁇ g/mL. Untreated cells were included as Control group for sample. Cells treated with Doxorubicin were included as Positive Control group. DMSO treated cells were included as control group for Doxorubicin. After treatment, cells were incubated for a time period of 24 h.
  • Annexin assay kit contains Annexin-V+7-AAD stain, which differentially labels apoptotic cells in different phases.
  • Four populations of cells can be determined from the flow cytometric plots as follows:
  • Annexin-V reagent 100 ⁇ l was added to each tube and incubated for 30 min at RT in dark.
  • Cells were counted using hemocytometer and plated in culture plates at density of 0.5 ⁇ 10 6 cells/well in DMEM+10% FBS in 6-well plates. Cells were incubated overnight to allow the cell recovery and exponential growth. Following overnight incubation, cells were sera starved in DMEM+1% FBS for 4 h. After 4 h, cells were treated with Test Items in DMEM+0% FBS at concentrations ranging from 2.5 ⁇ g/mL-80 ⁇ g/mL. Untreated cells were included as Control group for test item. Cells treated with Doxorubicin were included as Positive Control group. DMSO treated cells were included as control group for Doxorubicin. After treatment, cells were incubated for a time period of 24 h.
  • Ethanol fixed cells were centrifuged at 450 g for 5 min, RT (low brake). The supernatant was carefully removed (not to touch the pellet) and discarded. (The pellet might not be visible but makes a thin film on surface of tube). 1 mL of 1 ⁇ PBS was added into pellet and resuspended gently. Cells were incubated for 1 min at RT. Cells were centrifuged at 450 g for 5 min, RT (low brake) (washing step). The supernatant was removed carefully leaving behind approx 20 ⁇ L-50 ⁇ L of PBS. 200 ⁇ L of Cell Cycle reagent was added into each tube.
  • Results for apoptotic effect of Recombinant lectin of SEQ ID NO 2 on MDA-MB-231 cell line demonstrated that it induced an increase in early, late apoptotic and necrotic cells. Additionally, enhancement of apoptotic (Sub G0/G1) cell population was also observed upon treatment of cells with SEQ ID NO. 2.
  • the cell line used for the study was EA.hy926 (Human endothelial cells) which was procured from National Centre for Cell Science, Pune (India).
  • the cell line was maintained in DMEM+10% FBS (heat inactivated) at 37° C. with 5% CO2, and 95% humidity.
  • the cells were counted using hemocytometer and plated in 96 well plates at the density of 5 ⁇ 10 3 cells/well/180 ⁇ l of the growth medium. Following overnight incubation, cells were treated with recombinant lectin having amino acid sequence of SEQ ID NO.2 at concentrations ranging from about 2.5-100 ⁇ g/mL.
  • Untreated cells with complete medium (10% FBS) served as complete medium control
  • cells with sera free medium served as SFM control
  • the cells treated with Paclitaxel served as positive control.
  • Photomicrographs of the scratch were taken at 24 h-72 h time points.
  • the photomicrographs obtained in the above step were analyzed for quantitative assessment of area of wound closure using ImageJ tool software. Percentage migration with respect to untreated cells at different time points was calculated.
  • VEGF secretion was studied in MDA-MB-231 (Human breast adenocarcinoma) and PANC-1 (Human pancreatic epitheloid carcinoma) which were procured from National Centre for Cell Science, Pune (India).
  • the cell lines were maintained in DMEM+10% PBS (heat inactivated) at 37° C. with 5% CO2, and 95% humidity.
  • Cells were counted and plated in 6-well plates for 24 h at a density of 0.5 ⁇ 10 6 cells/well. The cells were incubated overnight under growth conditions as described above so as to allow cell recovery and exponential growth. Cells were treated with each test item at different concentrations for 24h in serum free media. Doxorubicin was used as positive control.
  • Secreted levels of VEGF were determined in supernatants after 24 hours using Human VEGF ELISA kit (R&D systems) as per manufacturer's protocol. Change in VEGF levels corresponding to each treatment was calculated using the following formula:
  • the recombinant lectin having the amino acid sequence of SEQ ID NO.2 was observed to increase the level of VEGF in MDA-MB-231 cells at most concentrations tested across the range of 2.5-80 ⁇ g/mL, compared to the untreated control (Table 14). An increase in VEGF levels in the presence of the lectin was also observed for PANC-1 cells at all concentrations tested across the same concentration range (Table 15).
  • angiogenesis-inducing compounds such as bFGF was introduced into cold liquid Matrigel which, after subcutaneous injection, solidified and permits penetration by host cells and the formation of new blood vessels (neovascularization).
  • Tris Buffered Saline TBS
  • the dose volume given to animal was 10 mL/Kg.
  • Group G3 (Sunitinib, 55 mg/Kg) showed maximum reduction i.e. 59.2% in hemoglobin content in homogenate of Matrigel plug whereas Group G4 (recombinant lectin having amino acid sequence of SEQ ID No 2, 10 mg/Kg; qdx15) showed 23.6% reduction in hemoglobin content when compared to positive control (Table 17).
  • Hemoglobin content in Matrigel plug and neovascularization of Matrigel plug histology indicate that recombinant lectin having amino acid sequence of SEQ ID NO. 2 (10 mg/Kg; qdx15) showed anti-angiogenic activity when compared to positive control group using Matrigel plug assay in C57BL/6 mice.
  • the in-vitro cytotoxic effects of recombinant lectin having amino acid sequence of SEQ ID NO. 2 was studied in a panel of Brain Tumor Cell lines consisting of Human Glioblastoma: LN-18, Human Glioblastoma: U251MG; Human Neuroblastoma: SH-SY-5Y; Human Meningioma: IOMM-Lee; Human Astrocytoma: U87MG; Rat C6 (Glioma) by MTT assay.
  • the SEQ ID NO. 2 was provided as aqueous solution (12.17 mg/mL) and the stock solution of SEQ ID NO. 2 was diluted in serum free medium (SFM) at different concentrations corresponding to 10 fold high concentration.
  • SFM serum free medium
  • the evaluation of pro-apoptotic effect of SEQ ID NO. 2 in brain tumor cell lines U251MG & IOMM Lee was conducted.
  • the cells were treated with SEQ ID NO. 2 at various concentrations around IC50 values.
  • Resultant effect on apoptosis was determined via externalization of Phosphatidyl Serine (PS) on cell membrane by Annexin-V staining, mitochondrial membrane depolarization by JC-1 staining and cell cycle distribution analysis by Propidium Iodide (PI) staining.
  • PS Phosphatidyl Serine
  • JC-1 mitochondrial membrane depolarization
  • PI Propidium Iodide
  • Increase in apoptotic markers in U251MG & IOMM-Lee cell lines reflected the pro-apoptotic potential of SEQ ID NO. 2 in brain tumor.
  • SEQ ID NO. 2 resulted in significant inhibition (p ⁇ 0.01, p ⁇ 0.001) of biomarkers (VEGF, VEGFR2, HGF, HGFR/c-MET, PDGF-BB, Notch-1) as compared to untreated control.
  • biomarkers VEGF, VEGFR2, HGF, HGFR/c-MET, PDGF-BB, Notch-1
  • TNF- ⁇ and Contactin-1 levels were also inhibited by SEQ ID NO. 2.
  • the purified recombinant lectin having amino acid sequence of SEQ ID NO. 2 was studied for its anti-cancer potential in different cell lines.
  • the anti-cancer potential of recombinant lectin having amino acid sequence of SEQ ID 2 was evaluated in-vitro on 13 different cancer cell lines and 3 normal cells using Calcein AM assay.
  • the cells were trypsinized, counted by Trypan blue method in Neuebauer's Chamber and plated in wells of flat bottom 96-well plate (dark walled plate, flat bottom) at the density corresponding to 10 ⁇ 10 3 cells/well/180 ⁇ l medium.
  • test item (20 ⁇ L) at concentrations ranging from 2.5-80 ⁇ g/mL, so that the total volume in each well is 200 ⁇ L.
  • the cells corresponding to positive control group were treated with Doxorubicin.
  • the untreated cells served as negative control that does not receive any treatment.
  • the cells were incubated with the test items or positive control for a time period of 48h. Following incubation, the cell cytotoxicity was estimated using Calcein AM Cell Viability Assay Kit from R&D systems (Cat No. 4892-010-K).
  • Percentage cytotoxicity wrt untreated cells was calculated using formula:
  • IC50 value was calculated using Graph-Pad Prism version 4.01 software.
  • the purified recombinant lectin having amino acid sequence of SEQ ID NO. 2 was studied for its anti-cancer potential in different cell lines.
  • the anti-cancer potential of recombinant lectin having amino acid sequence of SEQ ID 2 was evaluated in-vitro on 7 different cancer cell lines and one normal cell line using MTT assay.
  • Doxorubicin was used as positive control.
  • the stock solutions of recombinant lectin having amino acid sequence of SEQ ID 2 were prepared in DMSO and used at final concentrations of 2.5 ⁇ g/mL, 5 ⁇ g/mL, 10 ⁇ g/mL, 20 ⁇ g/mL, 40 ⁇ g/mL and 80 ⁇ g/mL.
  • stock solution of Doxorubicin was prepared in DMSO and used at final concentration of 0.1 ⁇ M, 1 ⁇ M, 10 ⁇ M, 25 ⁇ M, 50 ⁇ M and 75 ⁇ M.
  • the cells were trypsinized, counted by Trypan blue method in Neuebauer's Chamber and plated in wells of flat bottom 96-well plate at the density corresponding to 10 ⁇ 10 3 cells/well/200 ⁇ L medium.
  • the media in the plate was replenished with 180 ⁇ L/well and then the cells were treated with 20 ⁇ L of each test item at concentrations ranging from 2.5 ⁇ g/mL-80 ⁇ g/mL in triplicates, so that the total volume in each well is 200 ⁇ L.
  • the cells corresponding to positive control group were treated with Doxorubicin.
  • the untreated cells served as negative control.
  • the cells were incubated with the test items or positive control for a time period of 48h. Following incubation, the cell cytotoxicity was estimated using MTT assay. Absorbance was measured at 540 nm. The percentage cytotoxicity corresponding to each treatment was calculated using the formula:
  • IC50 value was calculated using Graph-Pad Prism version 4.01 software.
  • SEQ ID NO. 2 showed cytotoxic effect on PANC-1 (Human pancreatic epitheloid carcinoma), MDA-MB-231 (Human breast adenocarcinoma) and T24 (Human Bladder Cancer) cell lines with IC 50 values of 24.3 ⁇ g/mL, 9.7 ⁇ g/mL, and 10.4 ⁇ g/mL respectively.
  • Recombinant lectin of SEQ ID NO. 2 showed IC50 value of >80 ⁇ g/ml for all other cell lines. The results are summarized in Table 24.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Immunology (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Botany (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Zoology (AREA)
  • Mycology (AREA)
  • Cell Biology (AREA)
  • Toxicology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
US17/923,732 2020-05-07 2021-04-28 Anti-cancer proteins Pending US20230348564A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
IN202021019406 2020-05-07
IN202021019406 2020-05-07
PCT/IB2021/053503 WO2021224724A1 (en) 2020-05-07 2021-04-28 Anti-cancer proteins

Publications (1)

Publication Number Publication Date
US20230348564A1 true US20230348564A1 (en) 2023-11-02

Family

ID=75914557

Family Applications (1)

Application Number Title Priority Date Filing Date
US17/923,732 Pending US20230348564A1 (en) 2020-05-07 2021-04-28 Anti-cancer proteins

Country Status (10)

Country Link
US (1) US20230348564A1 (ja)
EP (1) EP4146243A1 (ja)
JP (1) JP2023524151A (ja)
KR (1) KR20230008072A (ja)
CN (1) CN115515619A (ja)
AU (1) AU2021267196A1 (ja)
BR (1) BR112022022329A2 (ja)
CA (1) CA3177876A1 (ja)
MX (1) MX2022013849A (ja)
WO (1) WO2021224724A1 (ja)

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR100486784B1 (ko) 2001-05-22 2005-04-29 김하형 솔비나무로 부터 추출된 렉틴 단백질, 그의 제조방법 및용도
KR20030028855A (ko) 2001-10-04 2003-04-11 (주)바이오메디팜 렉틴으로 강화된 겨우살이 추출물을 유효성분으로 하는항암제용 조성물
KR100492940B1 (ko) 2002-05-27 2005-06-02 김종배 항종양활성 증진용 조성물
TR201901202T4 (tr) 2009-02-18 2019-02-21 Karnatak Univ Antitümör aktivitesi olan rekombinant lektinlere bağlanan kanser hücresi ve hazirlama yöntemi.
WO2014203261A2 (en) 2010-02-02 2014-12-24 Unichem Laboratories Ltd. Recombinant lectin and method of preparing thereof
EP2508195A1 (de) 2011-04-06 2012-10-10 Cytavis BioPharma GmbH Arzneimittel enthaltend rekombinante Mistellektine zur Behandlung des malignen Melanoms
WO2015081290A1 (en) 2013-11-27 2015-06-04 Inis Biotech Llc Methods for modulating angiogenesis of cancers refractory to anti-vegf treatment
CN106397554A (zh) 2016-10-19 2017-02-15 上海市农业科学院 一种蛹虫草凝集素蛋白Lectin‑ccm3及其制备方法和应用
SG11202101391WA (en) 2018-08-31 2021-03-30 Unichem Lab Ltd Recombinant lectin variants
CA3112250A1 (en) * 2018-09-16 2020-04-16 Unichem Laboratories Ltd Protein for treatment of inflammatory diseases

Also Published As

Publication number Publication date
JP2023524151A (ja) 2023-06-08
KR20230008072A (ko) 2023-01-13
EP4146243A1 (en) 2023-03-15
MX2022013849A (es) 2023-01-16
CA3177876A1 (en) 2021-11-11
AU2021267196A1 (en) 2023-01-05
BR112022022329A2 (pt) 2022-12-13
CN115515619A (zh) 2022-12-23
WO2021224724A1 (en) 2021-11-11

Similar Documents

Publication Publication Date Title
Mercurio et al. Targeting CXCR4 by a selective peptide antagonist modulates tumor microenvironment and microglia reactivity in a human glioblastoma model
Yakes et al. Cabozantinib (XL184), a novel MET and VEGFR2 inhibitor, simultaneously suppresses metastasis, angiogenesis, and tumor growth
Hamano et al. Thrombospondin-1 associated with tumor microenvironment contributes to low-dose cyclophosphamide-mediated endothelial cell apoptosis and tumor growth suppression
Feldman et al. Erythropoietin stimulates growth and STAT5 phosphorylation in human prostate epithelial and prostate cancer cells
Kohno et al. A novel hybrid peptide targeting EGFR-expressing cancers
Lang et al. Combating head and neck cancer metastases by targeting Src using multifunctional nanoparticle-based saracatinib
Ju et al. Octreotide-modified liposomes containing daunorubicin and dihydroartemisinin for treatment of invasive breast cancer
JP2009501237A (ja) 生物活性fus1ペプチドおよびナノ粒子−ポリペプチド複合体
Fu et al. Specific amino acid dependency regulates invasiveness and viability of androgen-independent prostate cancer cells
Hua et al. Improved Antiglioblastoma Activity and BBB Permeability by Conjugation of Paclitaxel to a Cell‐Penetrative MMP‐2‐Cleavable Peptide
Wang et al. Polyplex-mediated inhibition of chemokine receptor CXCR4 and chromatin-remodeling enzyme NCOA3 impedes pancreatic cancer progression and metastasis
Kwak et al. Synergistic antitumor effects of combination treatment with metronomic doxorubicin and VEGF-targeting RNAi nanoparticles
Yu et al. Polymeric chloroquine as an inhibitor of cancer cell migration and experimental lung metastasis
Lee et al. Tpl2 inhibitors thwart endothelial cell function in angiogenesis and peritoneal dissemination
Sun et al. Targeting epirubicin plus quinacrine liposomes modified with DSPE-PEG2000-C (RGDfK) conjugate for eliminating invasive breast cancer
Kim et al. The coffee diterpene kahweol inhibits metastasis by modulating expressions of MMPs and VEGF via STAT3 inactivation
ES2800162T3 (es) Uso terapéutico de moduladores funcionales negativos de la eritropoyetina
Shi et al. Saikosaponin-A exhibits antipancreatic cancer activity by targeting the EGFR/PI3K/Akt pathway
CN111032075A (zh) 用于治疗癌症的方法和组合物
Zhu et al. A new HDAC inhibitor cinnamoylphenazine shows antitumor activity in association with intensive macropinocytosis
Zhou et al. Antitumor effects of hyperthermic CO 2 pneumoperitoneum on human gastric cancer cells
US20230348564A1 (en) Anti-cancer proteins
US20060025485A1 (en) Hydroxybenazamide compounds for treatment of cancer
Hu et al. A nanometer-sized protease inhibitor for precise cancer diagnosis and treatment
Gao et al. Novel fusion protein PK5-RL-Gal-3C inhibits hepatocellular carcinoma via anti-angiogenesis and cytotoxicity

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION